Skip to main content

Role of albumin in the preservation of endothelial glycocalyx integrity and the microcirculation: a review

Abstract

The endothelial glycocalyx comprises a complex layer of membrane-bound proteoglycans, secreted glycosaminoglycans, glycoproteins, glycolipids and bound plasma proteins such as albumin and antithrombin associated with the endothelial surface. The glycocalyx plays an important role in vascular homeostasis, regulating vascular permeability and cell adhesion, and acts as a mechanosensor for hemodynamic shear stresses; it also has antithrombotic and anti-inflammatory functions. Plasma proteins such as albumin are physiologically bound within the glycocalyx, thus contributing to stability of the layer. Albumin is the major determinant of plasma colloid osmotic pressure. In addition, albumin transports sphingosine-1-phosphate which has protective endothelial effects, acts as a free radical scavenger, and has immunomodulatory and anti-inflammatory effects. This review examines the physiological function of the endothelial glycocalyx and the role of human albumin in preserving glycocalyx integrity and the microcirculation.

The endothelial glycocalyx

Composition and structure of the endothelial glycocalyx

The endothelial glycocalyx is a complex carbohydrate-rich gel-like layer lining the luminal surface of blood vessels [1]; it functions as a barrier between the blood and vessel wall [2, 3]. The glycocalyx layer is composed of membrane-bound proteoglycans, secreted glycosaminoglycans (GAGs), sialic acid-containing glycoproteins, and glycolipids associated with the endothelial surface (Fig. 1) [4]. The main proteoglycans of the endothelial glycocalyx are membrane-spanning syndecans and glycosylphosphatidylinositol-linked glypicans which carry the two main GAGs, heparan sulfate and chondroitin sulfate, through covalent attachment to the protein core. Syndecans carry both GAGs, while glypicans carry only heparan sulfate. Table 1 summarizes the characteristics of core proteoglycans in the glycocalyx [5]. A third major GAG, hyaluronan, is secreted by endothelial cells but is not covalently linked to a core protein; it binds to cell surface adhesion receptors such as CD44. Plasma proteins such as albumin and antithrombin are also bound within the glycocalyx [4, 6,7,8,9].

Fig. 1
figure 1

Structure of the endothelial glycocalyx illustrating proteoglycans and glycosaminoglycans. Reproduced with permission from [4]. GPI glycosylphosphatidylinositol

Table 1 Characterization of proteoglycan core proteins in the glycocalyx.

The term endothelial surface layer is sometimes used to describe the intimal surface of blood vessels comprising the endothelial glycocalyx and associated components derived from endothelial cells and plasma [3, 10]. The thickness of the glycocalyx/endothelial surface layer (Fig. 2) [11] varies depending on the method used for measurement [9]. Intravital microscopy and orthogonal polarization spectral imaging are techniques which indirectly measure the endothelial glycocalyx in vivo, and the glycocalyx can be measured indirectly ex vivo using microparticle image velocimetry. The glycocalyx can be measured directly in vitro using transmission electron microscopy, confocal laser scanning microscopy and atomic force microscopy, and ex vivo using two-photon laser scanning microscopy [12, 13]. In humans, the mean microvascular glycocalyx thickness estimated using orthogonal polarization spectral imaging was about 0.5 µm (range 0.3 to 0.75 µm) [14].

Fig. 2
figure 2

Reproduced with permission from [11]

Electron micrograph of a cross-sectional image of a coronary endothelial glycocalyx (courtesy of B. van den Berg, Maastricht University).

Physiological function of the endothelial glycocalyx

The outermost layer of the microvasculature serves as a regulatory barrier of vascular permeability. It participates in mechanotransduction by sensing fluid shear forces and regulating the vascular tone. The endothelial glycocalyx, which has an important role in maintaining vascular homeostasis [9], also has several anti-adhesive and antithrombotic effects on the surface of endothelial cells and can protect endothelial cells from oxidative stress [3, 6,7,8,9, 15, 16].

Regulation of vascular permeability and barrier function

Starling’s original model of transvascular fluid exchange, which depends on a balance between hydrostatic and oncotic pressure gradients in semi-permeable capillaries, fails to explain the clinical responses observed after fluid resuscitation. A revised Starling model, first proposed by Levick and Michel [17] which incorporates the effect of the endothelial glycocalyx, basement membrane and extracellular matrix on fluid exchange, provides a better explanation of fluid transvascular interchanges and a patient’s response to fluid resuscitation because the effect of the glycocalyx in reducing fluid extravasation was unknown. Consequently, in Starling’s original model, the observed extracellular volume distribution following fluid resuscitation was not predicted by the model [2, 16,17,18,19]. Starling’s original principle and the revised Starling equation and glycocalyx model are compared in Additional file 1: Table S1 [18] and shown in Fig. 3 [2]. The revised Starling model proposes that the endothelial glycocalyx is the key determinant of hydrostatic and oncotic pressure gradients between the capillary lumen and the interstitium. Important Starling forces are the transendothelial pressure difference (Pc − Pis) and colloid osmotic pressure difference between plasma and the subglycocalyx (πp − πsg). This oncotic pressure difference explains the failure of the interstitial protein concentration to influence fluid movement [2, 17, 18].

Fig. 3
figure 3

The revised Starling principle. Reproduced with permission from [2]. Jv/A filtered volume per unit area; Lp hydraulic conductance; Pc capillary hydrostatic pressure; Pis interstitial hydrostatic pressure; σ osmotic reflection co-efficient; πp oncotic pressure in the luminal side of endothelial surface layer; πsg oncotic pressure in the subglycocalyx space

Under physiological conditions, the glycocalyx acts as a barrier against the shift of albumin and other circulating plasma components (mainly other proteins) across the endothelium [20, 21].

Mechanosensory function

Mechanical forces on endothelial cells generated by blood flow evoke biochemical responses that modulate endothelial structure and function through a process known as mechanotransduction. The glycocalyx acts as a cytoskeleton for endothelial cells. Mechanical distortion of ‘bush-like’ clusters of proteoglycans projecting from anchor points in the endothelial cell cytoskeleton generates forces which can deform the cytoskeleton. An associated increase in the expression of endothelial nitric oxide (NO) synthase catalyzes the production of NO, dilating vessels and reducing stress [2, 22, 23].

Rheological function

The glycocalyx has a vasculo-protective role by repelling red blood cells and by physically inhibiting the interaction of endothelial cell adhesion molecules (e.g., integrins and members of the immunoglobulin superfamily) with circulating platelets and leukocytes [9, 10].

Anticoagulation function

Several important anticoagulant mediators bind to GAGs located in the glycocalyx. These include antithrombin which binds via heparan sulfate to inhibit thrombin and activated factors IX and X; heparin cofactor II which is activated by dermatan sulfate; and tissue factor pathway inhibitor which binds via heparan sulfate to inhibit factors VIIa and Xa. Thrombomodulin, an endogenous anticoagulant produced by endothelial cells, interacts with thrombin to activate the protein C anticoagulant pathway [2].

Protective function against free radicals

Glycocalyx binding of enzymes such as extracellular superoxide dismutase protects endothelial cells against oxidative stress from reactive oxygen species (ROS) while maintaining NO availability, thus preventing endothelial dysfunction [9, 24].

Physiological role of endogenous albumin

Although albumin has a net negative charge, its amphoteric nature promotes tight binding to the glycocalyx with the net effect of reducing hydraulic conductivity across the vascular barrier, resisting glycocalyx degradation (i.e., protecting against shedding) and thereby contributing to maintenance of vascular integrity and normal capillary permeability, and facilitating transmission of shear stress [2, 15, 23, 25].

Under physiological conditions, the concentration of intravascular albumin is the major determinant of plasma colloid osmotic pressure [18].

Exposed thiol groups on the albumin molecule act as a scavenger for ROS such as superoxide (O2) and hydroxyl (OH) radicals and reactive nitrogen species, e.g., peroxynitrite radicals. Albumin has an additional anti-oxidant effect through binding to free copper ions (Cu2+) which are known to accelerate the production of free radicals [26, 27].

Albumin also has immunomodulatory and anti-inflammatory effects through binding of bacterial products, modulation of antigen-presenting cell function, modulation of cytokine production, and reducing hypoxia-inducible factor-1α gene expression which is upregulated in response to low oxygen concentrations [25].

Along with lipoproteins, albumin has an important role in delivering sphingosine-1-phosphate (S1P) to the endothelial cell surface where it functions in maintaining normal vascular permeability [28]. S1P protects endothelial cells by suppressing the activity of metalloproteinases, stabilizes the glycocalyx by reducing GAG degradation and shedding [21, 22], and regulates barrier function by modulating the expression of vascular endothelial-cadherin and β-catenin at endothelial cell–cell contact regions [29].

Post-translational modifications of human serum albumin include glycation, cysteinylation, S-nitrosylation, S-guanylation and S-transnitrosation which can affect the binding of some exogenous drugs [30]. In addition, advanced glycation end (AGE)-modified albumin can induce proinflammatory signaling through activation of AGE receptors [31]. This was illustrated in a murine model of peritonitis and sepsis where administration of therapeutic infusion solutions containing high concentrations of AGE-modified albumin reduced survival [32].

Alterations of the endothelial glycocalyx

Pathologies/interventions associated with glycocalyx alterations

Glycocalyx and endothelial cell damage, or endotheliopathy as it is known [33], occur in several clinical situations including ischemia–reperfusion injury, hypoxia/reoxygenation, inflammation, sepsis, hemorrhagic shock, hypervolemia, hyperglycemia, excessive shear stress and coronary artery bypass surgery [23, 34]. These injuries determine pathological changes in the endothelial glycocalyx such as impaired mechanotransduction, increased egress of leukocytes, loss of coagulation control, loss of anti-oxidant defense, loss of deposited growth factors, and increased vascular permeability (Fig. 4) [10, 35]. In a clinical context, disruption of the endothelial glycocalyx layer can lead to development of interstitial edema in some patients, notably those with inflammatory conditions such as sepsis [36].

Fig. 4
figure 4

Pathologies/interventions associated with glycocalyx alterations. Reproduced with permission from [10]. ecSOD extracellular superoxide dismutase; MMP matrix metalloprotease; NO nitric oxide. Sulodexide is a highly purified glycosaminoglycan mixture of low molecular weight heparin plus dermatan sulfate [30]

Ischemia–reperfusion injury

Ischemia–reperfusion injury results in tissue damage following disruption of the glycocalyx [37]. Microvascular endothelial cell dysfunction produces organ dysfunction locally or systemically, including systemic inflammatory response syndrome [38]. Cardiac ischemia–reperfusion injury may occur during procedures such as coronary artery bypass grafting, percutaneous coronary angioplasty, and heart transplant surgery [39], with cardiac surgery per se also producing glycocalyx damage [40].

Sepsis

Glycocalyx shedding is a feature of inflammation, trauma and sepsis, and is mediated by pro-inflammatory agents including tumor necrosis factor-α, C-reactive protein, adenosine, bradykinin, histamine, platelet-activating factor, and bacterial lipopolysaccharide. Glycocalyx shedding leads to increased vascular permeability, tissue edema and relative hypovolemia [41,42,43].

Many features of endothelial dysfunction in sepsis are shared with aging and with numerous chronic diseases such as hypertension, dyslipidemia, diabetes mellitus (DM), cardiovascular disease (CVD), cerebrovascular disease, chronic kidney disease (CKD), chronic pulmonary disease, liver disease, and cancer. Common features include glycocalyx degradation and shedding; increased oxidative stress and systemic inflammation; intercellular junction disassembly, endothelial cell death and blood–tissue barrier disruption; enhanced leukocyte adhesion and extravasation; and induction of procoagulant and antifibrinolytic pathways [43].

Hemorrhagic shock

Endothelial glycocalyx shedding has been shown in rodent models of hemorrhagic shock, although the underlying mechanism is unknown [44,45,46]. Recent results in rats subjected to non-traumatic hemorrhagic shock showed glycocalyx degradation, which was independent of increased vascular barrier permeability [46].

Hyperglycemia

Evidence from rodent models [47] and clinical studies in volunteers [48] suggests that acute and chronic hyperglycemia can cause glycocalyx damage. The link between DM and CVD is well established with CVD being the most common cause of morbidity and mortality in diabetic patients [49]. A role for heparan sulfates in the development of widespread vascular endothelial damage leading to albuminuria and associated complications in patients with type 1 DM was suggested by Deckert and colleagues who formulated the ‘Steno hypothesis’ which proposes that albumin leakage results from extensive vascular damage [50]. Exposure of vascular endothelial cells to hyperglycemia and advanced glycosylation end products causes glycocalyx disintegration with increased leukocyte egress and release of human protease activated receptor 2 agonists, together with endothelial NO synthase uncoupling, resulting in reduced NO availability and increased vascular permeability [51].

Other

High-density lipoprotein cholesterol (HDL-C) may have a role as a causal contributor to sepsis survival [52, 53]. Low HDL-C levels have been shown to be a strong predictor of organ dysfunction or death in patients with suspected sepsis [54]. As HDL-C is able to bind and sequester pathogenic lipids (e.g., endotoxins), its modification might be a novel therapeutic strategy for treating sepsis [55].

Potential effects of fluid therapy

Hypervolemia and type of fluid administered

A direct comparison of the hemodynamic effects of resuscitation fluids showed that colloids significantly increased plasma volume, cardiac index, and central venous pressure compared with crystalloids (p < 0.05), despite infusion of a higher volume of crystalloids (median 1800 vs 1500 mL) [55]. This is relevant because the duration of fluid infusion in fluid challenge significantly influences fluid responsiveness [56]. Fluid responsiveness does not equate with fluid requirement. Excess volume in the circulation is compensated by redistribution between stressed and unstressed volume [57] and, in the worst cases, by a leak to the interstitial space worsening tissue oxygenation [58]. A recently reported randomized trial found no differences between a slow (180 min) or rapid (30 min) infusion of 5% albumin on plasma volume expansion in patients following major abdominal surgery, and the rapid infusion had no effect on vascular leak [59].

As the type of fluid infused has an important effect on the glycocalyx, differences between albumin and crystalloids on glycocalyx function have been described.

In in vivo experiments of anesthetized rats subjected to hemorrhagic shock followed by fluid resuscitation, administration of normal saline failed to restore endothelial glycocalyx thickness and plasma levels of syndecan-1 (indicating failure to repair the glycocalyx), leading to a marked increase in vascular permeability and leukocyte rolling/adhesion. In contrast, albumin stabilized permeability and leukocyte rolling/adhesion, effects which were comparable to fresh frozen plasma. Albumin partially restored endothelial glycocalyx thickness, and lowered plasma syndecan-1 to baseline levels [60].

A recent review of commonly used resuscitation fluids for use in the critically ill highlighted the need to consider not only the oncotic properties of a fluid, but also its relative ability to protect and restore the endothelial glycocalyx. In this regard, evidence from observational and pre-clinical in vitro and in vivo studies indicates that albumin and fresh frozen plasma are superior to crystalloids and artificial colloids [61], although prospective studies are needed to confirm these findings.

Markers of endothelial damage

Biomarkers of endothelial damage have been developed with most applied prognostically for conditions of systemic inflammation and sepsis [43, 62,63,64,65,66,67,68,69,70,71] (Box 1). In patients with septic shock, increased plasma angiopoietin-2 levels were associated with higher fluid overload, hepatic and coagulation dysfunction, acute kidney injury, mortality, and plasma cytokines, likely as the result of increased vascular leakage [72].

Assessment of glycocalyx damage using a variety of biomarkers has provided evidence of glycocalyx degradation in a range of clinical conditions including trauma, CKD, myeloid leukemia, acute decompensated heart failure, and Crohn’s disease [5].

Role of human albumin in maintaining glycocalyx integrity

Pre-clinical studies of the effect of human albumin on the glycocalyx and microcirculation

Pre-clinical studies which illustrate the mechanism of action of albumin, and its effects in models of hemorrhagic shock, endotoxemia, vascular permeability and ischemia are summarized in Table 2 [13, 14, 60, 73,74,75,76,77,78,79,80,81,82,83,84,85,86,87]. Results from in vitro, in vivo, and ex vivo experiments illustrate the multifunctional nature of albumin including maintaining glycocalyx integrity and partially restoring impaired vascular permeability via release of S1P from RBCs; anti-inflammatory and anti-oxidative effects; improvement of the microcirculation and hemodynamics following hemorrhagic shock or endotoxemia; and acting as an effective plasma volume expander.

Table 2 Preclinical models of albumin

Clinical studies on the effect of human albumin in the glycocalyx and microcirculation

A recent study of patients with septic shock (n = 30) reported that, compared with saline, albumin improved skin endothelial cell function, improving microcirculatory blood flow. These beneficial effects may be independent of the oncotic properties of albumin as neither cardiac output nor skin blood flow differed between albumin- and saline-treated patients [88].

Conclusions and expert opinion

The endothelial glycocalyx plays an important role in regulating vascular permeability. Glycocalyx and endothelial cell damage occurs in several clinical situations including sepsis, hemorrhagic shock, hypervolemia, and hyperglycemia. Albumin is physiologically bound within the glycocalyx, protecting against shedding and contributing to the maintenance of vascular integrity and normal capillary permeability. Owing to these properties, albumin has the potential to improve outcomes in clinical scenarios characterized by damaged glycocalyx. Based on our review and interpretation of the available literature, we provide an opinion on the most suitable applications for albumin and highlight areas which require additional research.

Monitoring the microcirculation and endothelial damage

Several techniques are available to monitor the microcirculation and endothelial damage. These include: intravital microscopy in in vivo animal models for visualization of vascular dynamic events such as microvascular permeability, vasotone and blood flow [89] or the glycocalyx [90, 91]; assessment of the microcirculation with potential to measure the glycocalyx in critically ill patients [92]; application of near-infrared spectroscopy to measure tissue oxygenation [93, 94]; measuring skin mottling over the anterior surface of the knee [94]; measuring microalbuminuria [95]; biomarkers of acute kidney injury [96]; measuring protein concentrations in alveolar fluid lavage; and hemostasis-related biomarkers, e.g., factor VIII, von Willebrand factor, International Normalized Ratio, partial thromboplastin time and platelet count.

Intravital microscopy using sidestream darkfield (SDF) imaging is a non-invasive method increasingly used to analyze the sublingual microcirculation. The technique visualizes erythrocytes within the microvasculature due to light emitted by a light emitting diode probe which is reflected by hemoglobin and detected by a SDF camera [97]. Total vessel density, perfused vessel density, proportion of perfused vessels and microvascular flow index are traditionally estimated by offline computer analysis although, more recently, point-of-care approaches using validated automatic software platforms have been described [98, 99]. SDF imaging detection of RBCs is used as a marker of microvascular perfusion, and measurement of the perfused boundary region (PBR) as an indirect marker for endothelial glycocalyx barrier dimensions. In a large study of overweight and obese individuals, the PBR and presence of RBCs in the microvascular circulation were markedly associated [100]. Hand-held intravital microscopy showed that sublingual microvascular blood flow alterations are common in patients with sepsis, with blood flow abnormality related to disease severity [92, 101]. Furthermore, sublingual microvascular glycocalyx is damaged in critically ill patients, especially those with sepsis [102, 103], but also after cardiac surgery with cardiopulmonary bypass [104, 105] and in emergency room and intensive care unit patients [106]. However, in patients with sepsis, there was no association of PBR and syndecan-1 values with established microcirculatory parameters [102], likely indicating that both alterations occur independently. These data should be treated with caution as the reproducibility of three sublingual microcirculation parameters (vascular density, RBC filling and PBR) estimated by SDF imaging is controversial and large studies are required to achieve statistically significant effects [107]. However, some studies have shown good reproducibility with the method if consecutive measurements are averaged [106, 108, 109]. The accuracy of in vivo glycocalyx measurement has been analyzed further with an in vitro approach using atomic force microscopy [102, 110]. Consensus European Society of Intensive Care Medicine guidelines provide 15 recommendations for acquisition and interpretation of microcirculatory images obtained with hand-held vital microscopes for assessment of the microcirculation in critically ill patients [92].

According to our clinical and scientific judgement, biomarkers of endothelial damage and/or evaluation of the sublingual microcirculation may have a role in identifying subgroups of patients at risk of morbidity and mortality.

We consider that albumin should be used in accepted indications in which it has a proven positive risk:benefit balance, mainly in septic patients (for initial resuscitation after adequate crystalloid infusion and hypoalbuminemic septic shock), for burn shock resuscitation and fluid maintenance, and in some cases of liver insufficiency. The benefits of albumin may relate to its ability to restore function to damaged glycocalyx, although further studies are required to confirm this relationship and identify thresholds for optimal benefit. The mechanism of action of these restorative effects also needs to be elucidated.

Further research

As the lung and kidney are the organs most affected by septic shock, pre-clinical investigation of the effects of albumin on permeability disorders of these organs should be conducted. The development of non-invasive imaging-based analysis tools to assess changes in permeability, diameter, and blood flow of vessels in response to specific stimuli is beginning to benefit research in both pre-clinical and clinical areas. It is likely that patients’ response to albumin may differ depending on localization of the blood vessel under investigation.

Detailed studies of the effect of albumin on oxidative stress are required and could be assessed using in vitro models (e.g., cultures of human endothelial cells and glycocalyx components), as these are more reproducible than pre-clinical animal studies.

The main objective of clinical studies of albumin should not be to evaluate its effect on overall mortality, but rather on more specific endpoints such as organ dysfunction. Investigating the economic impact of albumin and its long-term consequences is also advised. Studies should accrue biological information, i.e., justification for the selected objectives. Smaller studies with homogeneous populations are preferred to large multicenter studies of patients with clinical heterogeneity.

In summary, additional research needs to be conducted to clarify the role of albumin as a protector or restorer of damaged glycocalyx, with the aim of identifying clinical applications.

Availability of data and materials

Not applicable.

References

  1. Weinbaum S, Tarbell JM, Damiano ER. The structure and function of the endothelial glycocalyx layer. Annu Rev Biomed Eng. 2007;9:121–67. https://doi.org/10.1146/annurev.bioeng.9.060906.151959.

    Article  CAS  PubMed  Google Scholar 

  2. Alphonsus CS, Rodseth RN. The endothelial glycocalyx: a review of the vascular barrier. Anaesthesia. 2014;69:777–84. https://doi.org/10.1111/anae.12661.

    Article  CAS  PubMed  Google Scholar 

  3. Sieve I, Münster-Kühnel AK, Hilfiker-Kleiner D. Regulation and function of endothelial glycocalyx layer in vascular diseases. Vascul Pharmacol. 2018;100:26–33. https://doi.org/10.1016/j.vph.2017.09.002.

    Article  CAS  PubMed  Google Scholar 

  4. Becker BF, Chappell D, Jacob M. Endothelial glycocalyx and coronary vascular permeability: the fringe benefit. Basic Res Cardiol. 2010;105:687–701. https://doi.org/10.1007/s00395-010-0118-z.

    Article  CAS  PubMed  Google Scholar 

  5. Ushiyama A, Kataoka H, Iijima T. Glycocalyx and its involvement in clinical pathophysiologies. J Intensive Care. 2016;4:59. https://doi.org/10.1186/s40560-016-0182-z.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Esko JD, Kimata K, Lindahl U. Chapter 16. Proteoglycans and sulfated glycosaminoglycans. In: Varki A, Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME, editors. Essentials of glycobiology. Cold Spring Harbor: Cold Spring Harbor Laboratory Press; 2009.

    Google Scholar 

  7. Broekhuizen LN, Mooij HL, Kastelein JJ, Stroes ES, Vink H, Nieuwdorp M. Endothelial glycocalyx as potential diagnostic and therapeutic target in cardiovascular disease. Curr Opin Lipidol. 2009;20:57–62. https://doi.org/10.1097/MOL.0b013e328321b587.

    Article  CAS  PubMed  Google Scholar 

  8. Chignalia AZ, Yetimakman F, Christiaans SC, Unal S, Bayrakci B, Wagener BM, Russell RT, Kerby JD, Pittet JF, Dull RO. The glycocalyx and trauma: a review. Shock. 2016;45:338–48. https://doi.org/10.1097/SHK.0000000000000513.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Iba T, Levy JH. Derangement of the endothelial glycocalyx in sepsis. J Thromb Haemost. 2019;17:283–94. https://doi.org/10.1111/jth.14371.

    Article  CAS  PubMed  Google Scholar 

  10. Song JW, Goligorsky MS. Perioperative implication of the endothelial glycocalyx. Korean J Anesthesiol. 2018;71:92–102. https://doi.org/10.4097/kjae.2018.71.2.92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lekakis J, Abraham P, Balbarini A, Blann A, Boulanger CM, Cockcroft J, Cosentino F, Deanfield J, Gallino A, Ikonomidis I, Kremastinos D, Landmesser U, Protogerou A, Stefanadis C, Tousoulis D, Vassalli G, Vink H, Werner N, Wilkinson I, Vlachopoulos C. Methods for evaluating endothelial function: a position statement from the European Society of Cardiology Working Group on Peripheral Circulation. Eur J Cardiovasc Prev Rehabil. 2011;18:775–89. https://doi.org/10.1177/1741826711398179.

    Article  PubMed  Google Scholar 

  12. Kang H, Deng X. The endothelial glycocalyx: visualization and measurement. J Biomed. 2017;2:120–3. https://doi.org/10.7150/jbm.20986.

    Article  Google Scholar 

  13. Job KM, O’Callaghan R, Hlady V, Barabanova A, Dull RO. The biomechanical effects of resuscitation colloids on the compromised lung endothelial glycocalyx. Anesth Analg. 2016;123:382–93. https://doi.org/10.1213/ANE.0000000000001284.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Nieuwdorp M, Meuwese MC, Mooij HL, Ince C, Broekhuizen LN, Kastelein JJ, Stroes ES, Vink H. Measuring endothelial glycocalyx dimensions in humans: a potential novel tool to monitor vascular vulnerability. J Appl Physiol. 1985;2008(104):845–52. https://doi.org/10.1152/japplphysiol.00440.2007.

    Article  Google Scholar 

  15. Becker BF, Chappell D, Bruegger D, Annecke T, Jacob M. Therapeutic strategies targeting the endothelial glycocalyx: acute deficits, but great potential. Cardiovasc Res. 2010;87:300–10. https://doi.org/10.1093/cvr/cvq137.

    Article  CAS  PubMed  Google Scholar 

  16. Uchimido R, Schmidt EP, Shapiro NI. The glycocalyx: a novel diagnostic and therapeutic target in sepsis. Crit Care. 2019;23:16. https://doi.org/10.1186/s13054-018-2292-6.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Levick JR, Michel CC. Microvascular fluid exchange and the revised Starling principle. Cardiovasc Res. 2010;87:198–210. https://doi.org/10.1093/cvr/cvq062.

    Article  CAS  PubMed  Google Scholar 

  18. Woodcock TE, Woodcock TM. Revised Starling equation and the glycocalyx model of transvascular fluid exchange: an improved paradigm for prescribing intravenous fluid therapy. Br J Anaesth. 2012;108:384–94. https://doi.org/10.1093/bja/aer515.

    Article  CAS  PubMed  Google Scholar 

  19. Thind GS, Zanders S, Baker JK. Recent advances in the understanding of endothelial barrier function and fluid therapy. Postgrad Med J. 2018;94:289–95. https://doi.org/10.1136/postgradmedj-2017-135125.

    Article  CAS  PubMed  Google Scholar 

  20. Rabelink TJ, de Zeeuw D. The glycocalyx–linking albuminuria with renal and cardiovascular disease. Nat Rev Nephrol. 2015;11:667–76. https://doi.org/10.1038/nrneph.2015.162.

    Article  CAS  PubMed  Google Scholar 

  21. Curry FE, Adamson RH. Endothelial glycocalyx: permeability barrier and mechanosensor. Ann Biomed Eng. 2012;40:828–39. https://doi.org/10.1007/s10439-011-0429-8.

    Article  CAS  PubMed  Google Scholar 

  22. Tarbell JM, Simon SI, Curry FR. Mechanosensing at the vascular interface. Annu Rev Biomed Eng. 2014;16:505–32. https://doi.org/10.1146/annurev-bioeng-071813-104908.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pillinger NL, Kam P. Endothelial glycocalyx: basic science and clinical implications. Anaesth Intensive Care. 2017;45:295–307. https://doi.org/10.1177/0310057X1704500305.

    Article  CAS  PubMed  Google Scholar 

  24. Reitsma S, Slaaf DW, Vink H, van Zandvoort MA, Oude Egbrink MG. The endothelial glycocalyx: composition, functions, and visualization. Pflugers Arch. 2007;454:345–59. https://doi.org/10.1007/s00424-007-0212-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ferrer R, Mateu X, Maseda E, Yébenes JC, Aldecoa C, De Haro C, Ruiz-Rodriguez JC, Garnacho-Montero J. Non-oncotic properties of albumin. A multidisciplinary vision about the implications for critically ill patients. Expert Rev Clin Pharmacol. 2018;11:125–37. https://doi.org/10.1080/17512433.2018.1412827.

    Article  CAS  PubMed  Google Scholar 

  26. Evans TW. Review article: albumin as a drug–biological effects of albumin unrelated to oncotic pressure. Aliment Pharmacol Ther. 2002;16(Suppl 5):6–11. https://doi.org/10.1046/j.1365-2036.2002.00190.x.

    Article  CAS  PubMed  Google Scholar 

  27. Taverna M, Marie AL, Mira JP, Guidet B. Specific antioxidant properties of human serum albumin. Ann Intensive Care. 2013;3:4. https://doi.org/10.1186/2110-5820-3-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Thuy AV, Reimann CM, Hemdan NY, Gräler MH. Sphingosine 1-phosphate in blood: function, metabolism, and fate. Cell Physiol Biochem. 2014;34:158–71. https://doi.org/10.1159/000362992.

    Article  CAS  PubMed  Google Scholar 

  29. Xiong Y, Hla T. S1P control of endothelial integrity. Curr Top Microbiol Immunol. 2014;378:85–105. https://doi.org/10.1007/978-3-319-05879-5_4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee P, Wu X. Review: modifications of human serum albumin and their binding effect. Curr Pharm Des. 2015;21(14):1862–5. https://doi.org/10.2174/1381612821666150302115025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Vetter SW. Glycated serum albumin and AGE receptors. Adv Clin Chem. 2015;72:205–75. https://doi.org/10.1016/bs.acc.2015.07.005.

    Article  CAS  PubMed  Google Scholar 

  32. Humpert PM, Lukic IK, Thorpe SR, Hofer S, Awad EM, Andrassy M, Deemer EK, Kasper M, Schleicher E, Schwaninger M, Weigand MA, Nawroth PP, Bierhaus A. AGE-modified albumin containing infusion solutions boosts septicaemia and inflammation in experimental peritonitis. J Leukoc Biol. 2009;86:589–97. https://doi.org/10.1189/jlb.1008646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Johansson PI, Stensballe J, Ostrowski SR. Shock induced endotheliopathy (SHINE) in acute critical illness—a unifying pathophysiologic mechanism. Crit Care. 2017;21:25. https://doi.org/10.1186/s13054-017-1605-5.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Doherty M, Buggy DJ. Intraoperative fluids: how much is too much? Br J Anaesth. 2012;109:69–79. https://doi.org/10.1093/bja/aes171.

    Article  CAS  PubMed  Google Scholar 

  35. Lauver DA, Lucchesi BR. Sulodexide: a renewed interest in this glycosaminoglycan. Cardiovasc Drug Rev. 2006;24:214–26. https://doi.org/10.1111/j.1527-3466.2006.00214.x.

    Article  CAS  PubMed  Google Scholar 

  36. Myburgh JA, Mythen MG. Resuscitation fluids. N Engl J Med. 2013;369:1243–51. https://doi.org/10.1056/NEJMoa1209759.

    Article  CAS  PubMed  Google Scholar 

  37. Seal JB, Gewertz BL. Vascular dysfunction in ischemia-reperfusion injury. Ann Vasc Surg. 2005;19:572–84. https://doi.org/10.1007/s10016-005-4616-7.

    Article  PubMed  Google Scholar 

  38. Carden DL, Granger DN. Pathophysiology of ischaemia-reperfusion injury. J Pathol. 2000;190:255–66. https://doi.org/10.1002/(SICI)1096-9896(200002)190:3%3c255:AID-PATH526%3e3.0.CO;2-6.

    Article  CAS  PubMed  Google Scholar 

  39. Reffelmann T, Kloner RA. The no-reflow phenomenon: a basic mechanism of myocardial ischemia and reperfusion. Basic Res Cardiol. 2006;101:359–72. https://doi.org/10.1007/s00395-006-0615-2.

    Article  PubMed  Google Scholar 

  40. Bruegger D, Rehm M, Abicht J, Paul JO, Stoeckelhuber M, Pfirrmann M, Reichart B, Becker BF, Christ F. Shedding of the endothelial glycocalyx during cardiac surgery: on-pump versus off-pump coronary artery bypass graft surgery. J Thorac Cardiovasc Surg. 2009;138:1445–7. https://doi.org/10.1016/j.jtcvs.2008.07.063.

    Article  PubMed  Google Scholar 

  41. Henry CB, Duling BR. TNF-alpha increases entry of macromolecules into luminal endothelial cell glycocalyx. Am J Physiol Heart Circ Physiol. 2000;279:2815–23. https://doi.org/10.1152/ajpheart.2000.279.6.H2815.

    Article  Google Scholar 

  42. Chappell D, Westphal M, Jacob M. The impact of the glycocalyx on microcirculatory oxygen distribution in critical illness. Curr Opin Anaesthesiol. 2009;22:155–62. https://doi.org/10.1097/ACO.0b013e328328d1b6.

    Article  PubMed  Google Scholar 

  43. Bermejo-Martin JF, Martín-Fernandez M, López-Mestanza C, Duque P, Almansa R. Shared features of endothelial dysfunction between sepsis and its preceding risk factors (aging and chronic disease). J Clin Med. 2018. https://doi.org/10.3390/jcm7110400.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Kozar RA, Peng Z, Zhang R, Holcomb JB, Pati S, Park P, Ko TC, Paredes A. Plasma restoration of endothelial glycocalyx in a rodent model of hemorrhagic shock. Anesth Analg. 2011;112:1289–95. https://doi.org/10.1213/ANE.0b013e318210385c.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Torres Filho I, Torres LN, Sondeen JL, Polykratis IA, Dubick MA. In vivo evaluation of venular glycocalyx during hemorrhagic shock in rats using intravital microscopy. Microvasc Res. 2013;85:128–33. https://doi.org/10.1016/j.mvr.2012.11.005.

    Article  PubMed  Google Scholar 

  46. Guerci P, Ergin B, Uz Z, Ince Y, Westphal M, Heger M, Ince C. Glycocalyx degradation is independent of vascular barrier permeability increase in nontraumatic hemorrhagic shock in rats. Anesth Analg. 2019;129:598–607. https://doi.org/10.1213/ANE.0000000000003918.

    Article  CAS  PubMed  Google Scholar 

  47. Zuurbier CJ, Demirci C, Koeman A, Vink H, Ince C. Short-term hyperglycemia increases endothelial glycocalyx permeability and acutely decreases lineal density of capillaries with flowing red blood cells. J Appl Physiol. 2005;99:1471–6. https://doi.org/10.1152/japplphysiol.00436.2005.

    Article  PubMed  Google Scholar 

  48. Nieuwdorp M, van Haeften TW, Gouverneur MC, Mooij HL, van Lieshout MH, Levi M, Meijers JC, Holleman F, Hoekstra JB, Vink H, Kastelein JJ, Stroes ES. Loss of endothelial glycocalyx during acute hyperglycemia coincides with endothelial dysfunction and coagulation activation in vivo. Diabetes. 2006;55:480–6. https://doi.org/10.2337/diabetes.55.02.06.db05-1103.

    Article  CAS  PubMed  Google Scholar 

  49. Leon BM, Maddox TM. Diabetes and cardiovascular disease: epidemiology, biological mechanisms, treatment recommendations and future research. World J Diabetes. 2015;6:1246–58. https://doi.org/10.4239/wjd.v6.i13.1246.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Deckert T, Feldt-Rasmussen B, Borch-Johnsen K, Jensen T, Kofoed-Enevoldsen A. Albuminuria reflects widespread vascular damage. The Steno hypothesis. Diabetologia. 1989;32:219–26.

    Article  CAS  Google Scholar 

  51. Goligorsky MS. Vascular endothelium in diabetes. Am J Physiol Renal Physiol. 2017;312:F266–75. https://doi.org/10.1152/ajprenal.00473.2016.

    Article  CAS  PubMed  Google Scholar 

  52. Trinder M, Genga KR, Kong HJ, Blauw LL, Lo C, Li X, Cirstea M, Wang Y, Rensen PCN, Russell JA, Walley KR, Boyd JH, Brunham LR. Cholesteryl ester transfer protein influences high-density lipoprotein levels and survival in sepsis. Am J Respir Crit Care Med. 2019;199:854–62. https://doi.org/10.1164/rccm.201806-1157OC.

    Article  CAS  PubMed  Google Scholar 

  53. Jones TK, Wong HR, Meyer NJ. HDL Cholesterol: a “pathogen lipid sink” for sepsis? Am J Respir Crit Care Med. 2019;199:812–4. https://doi.org/10.1164/rccm.201811-2084ED.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Cirstea M, Walley KR, Russell JA, Brunham LR, Genga KR, Boyd JH. Decreased high-density lipoprotein cholesterol level is an early prognostic marker for organ dysfunction and death in patients with suspected sepsis. J Crit Care. 2017;38:289–94. https://doi.org/10.1016/j.jcrc.2016.11.041.

    Article  CAS  PubMed  Google Scholar 

  55. van der Heijden M, Verheij J, van Nieuw Amerongen GP, Groeneveld AB. Crystalloid or colloid fluid loading and pulmonary permeability, edema, and injury in septic and nonseptic critically ill patients with hypovolemia. Crit Care Med. 2009;37:1275–81. https://doi.org/10.1097/CCM.0b013e31819cedfd.

    Article  PubMed  Google Scholar 

  56. Toscani L, Aya HD, Antonakaki D, Bastoni D, Watson X, Arulkumaran N, Rhodes A, Cecconi M. What is the impact of the fluid challenge technique on diagnosis of fluid responsiveness? A systematic review and meta-analysis. Crit Care. 2017;21:207. https://doi.org/10.1186/s13054-017-1796-9.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Funk DJ, Jacobsohn E, Kumar A. The role of venous return in critical illness and shock-part I: physiology. Crit Care Med. 2013;41:255–62. https://doi.org/10.1097/CCM.0b013e3182772ab6.

    Article  PubMed  Google Scholar 

  58. Spiegel R. Stressed vs. unstressed volume and its relevance to critical care practitioners. Clin Exp Emerg Med. 2016;3:52–4. https://doi.org/10.15441/ceem.16.128.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Statkevicius S, Bonnevier J, Fisher J, Bark BP, Larsson E, Öberg CM, Tingstedt B, Bentzer P. Albumin infusion rate and plasma volume expansion: a randomized clinical trial in postoperative patients after major surgery. Crit Care. 2019;23:191. https://doi.org/10.1186/s13054-019-2477-7.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Torres LN, Chung KK, Salgado CL, Dubick MA, Torres Filho IP. Low-volume resuscitation with normal saline is associated with microvascular endothelial dysfunction after hemorrhage in rats, compared to colloids and balanced crystalloids. Crit Care. 2017;21:160. https://doi.org/10.1186/s13054-017-1745-7.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Milford EM, Reade MC. Resuscitation fluid choices to preserve the endothelial glycocalyx. Crit Care. 2019;23:77. https://doi.org/10.1186/s13054-019-2369-x.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Schmidt EP, Overdier KH, Sun X, Lin L, Liu X, Yang Y, Ammons LA, Hiller TD, Suflita MA, Yu Y, Chen Y, Zhang F, Cothren Burlew C, Edelstein CL, Douglas IS, Linhardt RJ. Urinary glycosaminoglycans predict outcomes in septic shock and acute respiratory distress syndrome. Am J Respir Crit Care Med. 2016;194:439–49. https://doi.org/10.1164/rccm.201511-2281OC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Hsiao SY, Kung CT, Tsai NW, Su CM, Huang CC, Lai YR, Wang HC, Cheng BC, Su YJ, Lin WC, Chiang YF, Lu CH. Concentration and value of endocan on outcome in adult patients after severe sepsis. Clin Chim Acta. 2018;483:275–80. https://doi.org/10.1016/j.cca.2018.05.007.

    Article  CAS  PubMed  Google Scholar 

  64. Ricciuto DR, dos Santos CC, Hawkes M, Toltl LJ, Conroy AL, Rajwans N, Lafferty EI, Cook DJ, Fox-Robichaud A, Kahnamoui K, Kain KC, Liaw PC, Liles WC. Angiopoietin-1 and angiopoietin-2 as clinically informative prognostic biomarkers of morbidity and mortality in severe sepsis. Crit Care Med. 2011;39:702–10. https://doi.org/10.1097/CCM.0b013e318206d285.

    Article  CAS  PubMed  Google Scholar 

  65. Mikacenic C, Hahn WO, Price BL, Harju-Baker S, Katz R, Kain KC, Himmelfarb J, Liles WC, Wurfel MM. Biomarkers of endothelial activation are associated with poor outcome in critical illness. PLoS ONE. 2015;10:e0141251. https://doi.org/10.1371/journal.pone.0141251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mikacenic C, Price BL, Harju-Baker S, O’Mahony DS, Robinson-Cohen C, Radella F, Hahn WO, Katz R, Christiani DC, Himmelfarb J, Liles WC, Wurfel MM. A two-biomarker model predicts mortality in the critically ill with sepsis. Am J Respir Crit Care Med. 2017;196:1004–11. https://doi.org/10.1164/rccm.201611-2307OC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Reilly JP, Wang F, Jones TK, Palakshappa JA, Anderson BJ, Shashaty MGS, Dunn TG, Johansson ED, Riley TR, Lim B, Abbott J, Ittner CAG, Cantu E, Lin X, Mikacenic C, Wurfel MM, Christiani DC, Calfee CS, Matthay MA, Christie JD, Feng R, Meyer NJ. Plasma angiopoietin-2 as a potential causal marker in sepsis-associated ARDS development: evidence from Mendelian randomization and mediation analysis. Intensive Care Med. 2018;44:1849–58. https://doi.org/10.1007/s00134-018-5328-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Paulus P, Jennewein C, Zacharowski K. Biomarkers of endothelial dysfunction: can they help us deciphering systemic inflammation and sepsis? Biomarkers. 2011;16(Suppl 1):S11–21. https://doi.org/10.3109/1354750X.2011.587893.

    Article  CAS  PubMed  Google Scholar 

  69. Andaluz-Ojeda D, Nguyen HB, Meunier-Beillard N, Cicuéndez R, Quenot JP, Calvo D, Dargent A, Zarca E, Andrés C, Nogales L, Eiros JM, Tamayo E, Bermejo-Martín JF, Charles PE. Superior accuracy of mid-regional proadrenomedullin for mortality prediction in sepsis with varying levels of illness severity. Ann Intensive Care. 2017;7:15. https://doi.org/10.1186/s13613-017-0238-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Walczak M, Suraj J, Kus K, Kij A, Zakrzewska A, Chlopicki S. Towards a comprehensive endothelial biomarkers profiling and endothelium-guided pharmacotherapy. Pharmacol Rep. 2015;67:771–7. https://doi.org/10.1016/j.pharep.2015.06.008.

    Article  CAS  PubMed  Google Scholar 

  71. Burger D, Touyz RM. Cellular biomarkers of endothelial health: microparticles, endothelial progenitor cells, and circulating endothelial cells. J Am Soc Hypertens. 2012;6:85–99. https://doi.org/10.1016/j.jash.2011.11.003.

    Article  CAS  PubMed  Google Scholar 

  72. Fisher J, Douglas JJ, Linder A, Boyd JH, Walley KR, Russell JA. Elevated plasma angiopoietin-2 levels are associated with fluid overload, organ dysfunction, and mortality in human septic shock. Crit Care Med. 2016;44:2018–27. https://doi.org/10.1097/CCM.0000000000001853.

    Article  CAS  PubMed  Google Scholar 

  73. Adamson RH, Clark JF, Radeva M, Kheirolomoom A, Ferrara KW, Curry FE. Albumin modulates S1P delivery from red blood cells in perfused microvessels: mechanism of the protein effect. Am J Physiol Heart Circ Physiol. 2014;306:H1011–7. https://doi.org/10.1152/ajpheart.00829.2013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Torres Filho IP, Torres LN, Salgado C, Dubick MA. Plasma syndecan-1 and heparan sulfate correlate with microvascular glycocalyx degradation in hemorrhaged rats after different resuscitation fluids. Am J Physiol Heart Circ Physiol. 2016;310:H1468–78. https://doi.org/10.1152/ajpheart.00006.2016.

    Article  PubMed  Google Scholar 

  75. Kremer H, Baron-Menguy C, Tesse A, Gallois Y, Mercat A, Henrion D, Andriantsitohaina R, Asfar P, Meziani F. Human serum albumin improves endothelial dysfunction and survival during experimental endotoxemia: concentration-dependent properties. Crit Care Med. 2011;39:1414–22. https://doi.org/10.1097/CCM.0b013e318211ff6e.

    Article  CAS  PubMed  Google Scholar 

  76. Lang JD Jr, Figueroa M, Chumley P, Aslan M, Hurt J, Tarpey MM, Alvarez B, Radi R, Freeman BA. Albumin and hydroxyethyl starch modulate oxidative inflammatory injury to vascular endothelium. Anesthesiology. 2004;100:51–8. https://doi.org/10.1097/00000542-200401000-00012.

    Article  CAS  PubMed  Google Scholar 

  77. Tatara T. The contribution of solute-solvent exchange at the membrane surface to the reduction by albumin of the hydraulic permeability coefficient of an artificial semipermeable membrane. Anesth Analg. 2003;97:1137–42. https://doi.org/10.1213/01.ANE.0000080612.68161.F0.

    Article  PubMed  Google Scholar 

  78. Wettstein R, Tsai AG, Erni D, Lukyanov AN, Torchilin VP, Intaglietta M. Improving microcirculation is more effective than substitution of red blood cells to correct metabolic disorder in experimental hemorrhagic shock. Shock. 2004;21:235–40. https://doi.org/10.1097/01.shk.0000114301.36496.ea.

    Article  PubMed  Google Scholar 

  79. Horstick G, Lauterbach M, Kempf T, Bhakdi S, Heimann A, Horstick M, Meyer J, Kempski O. Early albumin infusion improves global and local hemodynamics and reduces inflammatory response in hemorrhagic shock. Crit Care Med. 2002;30:851–5. https://doi.org/10.1097/00003246-200204000-00023.

    Article  CAS  PubMed  Google Scholar 

  80. Damiani E, Ince C, Orlando F, Pierpaoli E, Cirioni O, Giacometti A, Mocchegiani F, Pelaia P, Provinciali M, Donati A. Effects of the infusion of 4% or 20% human serum albumin on the skeletal muscle microcirculation in endotoxemic rats. PLoS ONE. 2016;11:e0151005. https://doi.org/10.1371/journal.pone.0151005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Cosenza L, Donati A, Cecero E, Carlucci M, Adembri C, Fusi F. Effects of albumin infusion on LPS-induced damage of mesenteric microcirculation: 12AP1-9. Eur J Anaesthesiol. 2012;29:176–7.

    Article  Google Scholar 

  82. Rehm M, Zahler S, Lötsch M, Welsch U, Conzen P, Jacob M, Becker BF. Endothelial glycocalyx as an additional barrier determining extravasation of 6% hydroxyethyl starch or 5% albumin solutions in the coronary vascular bed. Anesthesiology. 2004;100:1211–23. https://doi.org/10.1097/00000542-200405000-00025.

    Article  CAS  PubMed  Google Scholar 

  83. Bansch P, Statkevicius S, Bentzer P. Plasma volume expansion with 5% albumin compared to Ringer’s acetate during normal and increased microvascular permeability in the rat. Anesthesiology. 2014;121:817–24. https://doi.org/10.1097/ALN.0000000000000363.

    Article  PubMed  Google Scholar 

  84. Zausig YA, Chappell D, Becker BF, Potschka D, Busse H, Nixdorf K, Bitzinger D, Jacob B, Jacob M. The impact of crystalloidal and colloidal infusion preparations on coronary vascular integrity, interstitial oedema and cardiac performance in isolated hearts. Crit Care. 2013;17:R203. https://doi.org/10.1186/cc12898.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Dubniks M, Persson J, Grände PO. Plasma volume expansion of 5% albumin, 4% gelatin, 6% HES 130/0.4, and normal saline under increased microvascular permeability in the rat. Intensive Care Med. 2007;33:293–9. https://doi.org/10.1007/s00134-006-0454-5.

    Article  CAS  PubMed  Google Scholar 

  86. Jacob M, Bruegger D, Rehm M, Welsch U, Conzen P, Becker BF. Contrasting effects of colloid and crystalloid resuscitation fluids on cardiac vascular permeability. Anesthesiology. 2006;104:1223–31. https://doi.org/10.1097/00000542-200606000-00018.

    Article  CAS  PubMed  Google Scholar 

  87. Belayev L, Pinard E, Nallet H, Seylaz J, Liu Y, Riyamongkol P, Zhao W, Busto R, Ginsberg MD. Albumin therapy of transient focal cerebral ischemia: in vivo analysis of dynamic microvascular responses. Stroke. 2002;33:1077–84. https://doi.org/10.1161/hs0402.105555.

    Article  PubMed  Google Scholar 

  88. Hariri G, Joffre J, Deryckere S, Bigé N, Dumas G, Baudel JL, Maury E, Guidet B, Ait-Oufella H. Albumin infusion improves endothelial function in septic shock patients: a pilot study. Intensive Care Med. 2018;44:669–71. https://doi.org/10.1007/s00134-018-5075-2.

    Article  CAS  PubMed  Google Scholar 

  89. Honkura N, Richards M, Laviña B, Sáinz-Jaspeado M, Betsholtz C, Claesson-Welsh L. Intravital imaging-based analysis tools for vessel identification and assessment of concurrent dynamic vascular events. Nat Commun. 2018;9:2746. https://doi.org/10.1038/s41467-018-04929-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Margraf A, Herter JM, Kühne K, Stadtmann A, Ermert T, Wenk M, Meersch M, Van Aken H, Zarbock A, Rossaint J. 6% Hydroxyethyl starch (HES 130/0.4) diminishes glycocalyx degradation and decreases vascular permeability during systemic and pulmonary inflammation in mice. Crit Care. 2018;22(1):111. https://doi.org/10.1186/s13054-017-1846-3.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Lukasz A, Hillgruber C, Oberleithner H, Kusche-Vihrog K, Pavenstädt H, Rovas A, Hesse B, Goerge T, Kümpers P. Endothelial glycocalyx breakdown is mediated by angiopoietin-2. Cardiovasc Res. 2017;113(6):671–80. https://doi.org/10.1093/cvr/cvx023.

    Article  CAS  PubMed  Google Scholar 

  92. Ince C, Boerma EC, Cecconi M, De Backer D, Shapiro NI, Duranteau J, Pinsky MR, Artigas A, Teboul JL, Reiss IKM, Aldecoa C, Hutchings SD, Donati A, Maggiorini M, Taccone FS, Hernandez G, Payen D, Tibboel D, Martin DS, Zarbock A, Monnet X, Dubin A, Bakker J, Vincent JL, Scheeren TWL. Cardiovascular Dynamics Section of the ESICM. Second consensus on the assessment of sublingual microcirculation in critically ill patients: results from a task force of the European Society of Intensive Care Medicine. Intensive Care Med. 2018;44:281–99. https://doi.org/10.1007/s00134-018-5070-7.

    Article  PubMed  Google Scholar 

  93. Boushel R, Langberg H, Olesen J, Gonzales-Alonzo J, Bülow J, Kjaer M. Monitoring tissue oxygen availability with near infrared spectroscopy (NIRS) in health and disease. Scand J Med Sci Sports. 2001;11:213–22. https://doi.org/10.1034/j.1600-0838.2001.110404.x.

    Article  CAS  PubMed  Google Scholar 

  94. Charlton M, Sims M, Coats T, Thompson JP. The microcirculation and its measurement in sepsis. J Intensive Care Soc. 2017;18:221–7. https://doi.org/10.1177/1751143716678638.

    Article  PubMed  Google Scholar 

  95. Redon J. Measurement of microalbuminuria–what the nephrologist should know. Nephrol Dial Transplant. 2006;21:573–6. https://doi.org/10.1093/ndt/gfk014.

    Article  PubMed  Google Scholar 

  96. Kashani K, Al-Khafaji A, Ardiles T, Artigas A, Bagshaw SM, Bell M, Bihorac A, Birkhahn R, Cely CM, Chawla LS, Davison DL, Feldkamp T, Forni LG, Gong MN, Gunnerson KJ, Haase M, Hackett J, Honore PM, Hoste EA, Joannes-Boyau O, Joannidis M, Kim P, Koyner JL, Laskowitz DT, Lissauer ME, Marx G, McCullough PA, Mullaney S, Ostermann M, Rimmelé T, Shapiro NI, Shaw AD, Shi J, Sprague AM, Vincent JL, Vinsonneau C, Wagner L, Walker MG, Wilkerson RG, Zacharowski K, Kellum JA. Discovery and validation of cell cycle arrest biomarkers in human acute kidney injury. Crit Care. 2013;17:R25. https://doi.org/10.1186/cc12503.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Goedhart PT, Khalilzada M, Bezemer R, Merza J, Ince C. Sidestream Dark Field (SDF) imaging: a novel stroboscopic LED ring-based imaging modality for clinical assessment of the microcirculation. Opt Express. 2007;15(23):15101–14. https://doi.org/10.1364/oe.15.015101.

    Article  CAS  PubMed  Google Scholar 

  98. Tanaka S, Harrois A, Nicolaï C, Flores M, Hamada S, Vicaut E, Duranteau J. Qualitative real-time analysis by nurses of sublingual microcirculation in intensive care unit: the MICRONURSE study. Crit Care. 2015;19:388. https://doi.org/10.1186/s13054-015-1106-3.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Naumann DN, Mellis C, Husheer SL, Hopkins P, Bishop J, Midwinter MJ, Hutchings SD. Real-time point of care microcirculatory assessment of shock: design, rationale and application of the point of care microcirculation (POEM) tool. Crit Care. 2016;20(1):310. https://doi.org/10.1186/s13054-016-1492-1.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Lee DH, Dane MJ, van den Berg BM, Boels MG, van Teeffelen JW, de Mutsert R, den Heijer M, Rosendaal FR, van der Vlag J, van Zonneveld AJ, Vink H, Rabelink TJ, NEO study group. Deeper penetration of erythrocytes into the endothelial glycocalyx is associated with impaired microvascular perfusion. PLoS ONE. 2014;9:e96477. https://doi.org/10.1371/journal.pone.0096477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. De Backer D, Creteur J, Preiser JC, Dubois MJ, Vincent JL. Microvascular blood flow is altered in patients with sepsis. Am J Respir Crit Care Med. 2002;166:98–104. https://doi.org/10.1164/rccm.200109-016oc.

    Article  PubMed  Google Scholar 

  102. Rovas A, Seidel LM, Vink H, Pohlkötter T, Pavenstädt H, Ertmer C, Hessler M, Kümpers P. Association of sublingual microcirculation parameters and endothelial glycocalyx dimensions in resuscitated sepsis. Crit Care. 2019;23:260. https://doi.org/10.1186/s13054-019-2542-2.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Donati A, Damiani E, Domizi R, Romano R, Adrario E, Pelaia P, Ince C, Singer M. Alteration of the sublingual microvascular glycocalyx in critically ill patients. Microvasc Res. 2013;90:86–9. https://doi.org/10.1016/j.mvr.2013.08.007.

    Article  PubMed  Google Scholar 

  104. Koning NJ, Vonk AB, Vink H, Boer C. Side-by-side alterations in glycocalyx thickness and perfused microvascular density during acute microcirculatory alterations in cardiac surgery. Microcirculation. 2016;23(1):69–74. https://doi.org/10.1111/micc.12260.

    Article  PubMed  Google Scholar 

  105. Dekker NAM, Veerhoek D, Koning NJ, van Leeuwen ALI, Elbers PWG, van den Brom CE, Vonk ABA, Boer C. Postoperative microcirculatory perfusion and endothelial glycocalyx shedding following cardiac surgery with cardiopulmonary bypass. Anaesthesia. 2019;74:609–18. https://doi.org/10.1111/anae.14577.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Rovas A, Lukasz AH, Vink H, Urban M, Sackarnd J, Pavenstädt H, Kümpers P. Bedside analysis of the sublingual microvascular glycocalyx in the emergency room and intensive care unit - the GlycoNurse study. Scand J Trauma Resusc Emerg Med. 2018;26:16. https://doi.org/10.1186/s13049-018-0483-4.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Valerio L, Peters RJ, Zwinderman AH, Pinto-Sietsma SJ. Reproducibility of sublingual microcirculation parameters obtained from sidestream darkfield imaging. PLoS ONE. 2019;14(3):e0213175. https://doi.org/10.1371/journal.pone.0213175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Weissgerber TL, Garcia-Valencia O, Milic NM, Codsi E, Cubro H, Nath MC, White WM, Nath KA, Garovic VD. Early onset preeclampsia is associated with glycocalyx degradation and reduced microvascular perfusion. J Am Heart Assoc. 2019;8(4):e010647. https://doi.org/10.1161/JAHA.118.010647.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Bol ME, Beurskens DMH, Delnoij TSR, Roekaerts PMHJ, Reutelingsperger CPM, Delhaas T, Van de Poll MCG, Sels JEM, Nicolaes GAF. Variability of microcirculatory measurements in critically ill patients. Shock. 2019. https://doi.org/10.1097/SHK.0000000000001470(Epub ahead of print).

    Article  Google Scholar 

  110. Drost CC, Rovas A, Kusche-Vihrog K, Van Slyke P, Kim H, Hoang VC, Maynes JT, Wennmann DO, Pavenstädt H, Linke W, Lukasz A, Hesse B, Kümpers P. Tie2 activation promotes protection and reconstitution of the endothelial glycocalyx in human sepsis. Thromb Haemost. 2019;119(11):1827–38. https://doi.org/10.1055/s-0039-1695768.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Writing assistance was provided by Rob Furlong and Kerry Dechant on behalf of Content Ed Net (Madrid, Spain) with funding from Grifols S.A. (Barcelona, Spain). Grifols is a manufacturer of albumin.

Funding

Funding received from Grifols.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to writing and editing the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Antonio Artigas.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Cesar Aldecoa has received payment for lectures and travel expenses from Fresenius, and Octapharma. Juan V Llau has received payment for lectures or participation in educational activities, or travel expenses from Cardinal, CSL-Behring, LFB, Octapharma, Rovi, Sanofi, and Vifor Pharma. Xavier Nuvials has received payment for lectures, and travel expenses from Astellas, Gilead, MSD, and Pfizer. Antonio Artigas has received research grants from Fisher & Paykel, Grifols, and Hill-Rom; has participated in scientific advisory boards for Global Sepsis Alliance, Grifols, and Lilly Foundation; has received payment for lectures or participation in educational activities from Astute, Edwards, and Grifols.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Additional file 1: Table S1.

Comparison of Starling’s original principle and the revised Starling equation and glycocalyx model. Adapted from [18].

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aldecoa, C., Llau, J.V., Nuvials, X. et al. Role of albumin in the preservation of endothelial glycocalyx integrity and the microcirculation: a review. Ann. Intensive Care 10, 85 (2020). https://doi.org/10.1186/s13613-020-00697-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13613-020-00697-1

Keywords